US20150238580A1 - Glycosidase regimen for treatment of infectious disease - Google Patents

Glycosidase regimen for treatment of infectious disease Download PDF

Info

Publication number
US20150238580A1
US20150238580A1 US14/431,249 US201314431249A US2015238580A1 US 20150238580 A1 US20150238580 A1 US 20150238580A1 US 201314431249 A US201314431249 A US 201314431249A US 2015238580 A1 US2015238580 A1 US 2015238580A1
Authority
US
United States
Prior art keywords
glycosidase
regimen
patient
infection
neuraminidase
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US14/431,249
Other languages
English (en)
Inventor
Ellis Kline
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Individual
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Priority to US14/431,249 priority Critical patent/US20150238580A1/en
Publication of US20150238580A1 publication Critical patent/US20150238580A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/12Viral antigens
    • A61K39/21Retroviridae, e.g. equine infectious anemia virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/47Hydrolases (3) acting on glycosyl compounds (3.2), e.g. cellulases, lactases
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • A61K9/006Oral mucosa, e.g. mucoadhesive forms, sublingual droplets; Buccal patches or films; Buccal sprays
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/02Nasal agents, e.g. decongestants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/04Drugs for disorders of the respiratory system for throat disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/08Bronchodilators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/02Drugs for disorders of the urinary system of urine or of the urinary tract, e.g. urine acidifiers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/16Otologicals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • A61P31/06Antibacterial agents for tuberculosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • A61P31/22Antivirals for DNA viruses for herpes viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • A61P33/06Antimalarials
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y302/00Hydrolases acting on glycosyl compounds, i.e. glycosylases (3.2)
    • C12Y302/01Glycosidases, i.e. enzymes hydrolysing O- and S-glycosyl compounds (3.2.1)
    • C12Y302/01018Exo-alpha-sialidase (3.2.1.18), i.e. trans-sialidase
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y302/00Hydrolases acting on glycosyl compounds, i.e. glycosylases (3.2)
    • C12Y302/01Glycosidases, i.e. enzymes hydrolysing O- and S-glycosyl compounds (3.2.1)
    • C12Y302/01129Endo-alpha-sialidase (3.2.1.129)
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to treatment and prevention of disease, and particularly the prevention, treatment and management of infectious disease, including chronic and non-chronic viral infections and other infectious disease with a glycosidase therapy.
  • chemotherapeutic, monoclonal, or cytokine therapies can result in pathogen resistance, toxic effects for the patient including immune suppression, loss of effectiveness over time, and can be cost prohibitive for many patients.
  • antiretroviral (ARV) therapy although successful in slowing the progression of AIDS, has transformed the disease into a chronic disease requiring long term treatment, and a basic acceptance of the very significant side effects and enormous cost of the drugs. Atun and Bateringaya, Building a during response to HIV/AIDS: implications for health systems, J.
  • the invention provides compositions and methods for treating or preventing infectious disease, including chronic infections, and highly contagious infectious agents that present an ongoing challenge for the immune system or public health generally.
  • the compositions and treatment regimens described herein may find use with other antiviral or antimicrobial therapies, as well as in conjunction with vaccination (e.g., non-adjuvant-containing vaccination) to boost vaccine effectiveness and/or extend the duration of protective effect.
  • the treatment comprises in vivo administration of a glycosidase enzyme regimen (e.g., a regimen of one or more glycosidase enzymes) to the patient.
  • the glycosidase regimen is not targeted by the patient's immune system.
  • the glycosidase regimen provides one or more glycosidase enzymes active for removal of one or more terminal glycosyl groups on mammalian cells (e.g., immune cells), infected cells, or other glycosylated targets leading to immune activation.
  • the glycosidase therapy elicits immune signaling cascades via its action on immune cells.
  • Targeted terminal glycosyl groups may comprise, for example, beta-galactosyl, N-acetylgalactosamino, fucosyl, glucosyl, N-acetylglucosamino, and mannosyl residues, among others.
  • the glycosidase regimen can include, in various embodiments, one or more of neuraminidase, galactosidase, N-acetylgalactosidase, fucosidase, glucosidase, N-acetylglucosaminidase, and mannosidase, among others.
  • the regimen increases immune signaling by removing effective amounts of glycosyl structures (e.g., sialic acid) from the surface of immune cells, infected cells, and/or other glycosylated targets.
  • the glycosidase regimen orchestrates or programs an effective immune response, allowing antigenic targeting of infected cells as well as eliciting proper levels of cytokine/chemokine cascades for therapy.
  • the glycosidase enzymes include at least one enzyme specific for a prominent terminal glycosyl residue (e.g., neuraminidase and/or galactosidase), and at least one enzyme specific for a prominent penultimate glycosyl residue (e.g., beta-galactosidase, fucosidase, or mannosidase) on the surface of immune cells.
  • a prominent terminal glycosyl residue e.g., neuraminidase and/or galactosidase
  • a prominent penultimate glycosyl residue e.g., beta-galactosidase, fucosidase, or mannosidase
  • such enzymes act synergistically with neuraminidase.
  • the regimen does not dysregulate (but instead coordinates) the patient's immune system, which is crucial in fighting infectious disease, and is effective even in the presence of certain levels of cytotoxic chemotherapies, which can have deleterious effects on immune cells. Further, the regimen is applicable for chronic therapy, or repeated therapy, since the agent(s) are not targeted by the immune system in various embodiments.
  • the regimen in various embodiments avoids excess removal of sialic acids or other glycosyl structures from normal cells so that they retain normal function.
  • the glycosidase regimen described herein reduces or eliminates the need for administration of other traditional antiviral or antimicrobial therapies.
  • the invention finds use in immunocompromised or immunosuppressed patients for increasing immune function.
  • the glycosidase regimen in various embodiments is not immune targeted, and thus the resulting glycosidase signaling can be used for long term therapy.
  • the invention provides methods for treating patients having a chronic viral infection through a non-acute regimen of a composition comprising a glycosidase formulation that is both tolerated by the immune system (“immune tolerant”) and sufficient for stimulating coordinated immune signaling.
  • a composition comprising a glycosidase formulation that is both tolerated by the immune system (“immune tolerant”) and sufficient for stimulating coordinated immune signaling.
  • the glycosidase composition thus provides for immune stimulation, such as through one or more integral immune modulation cascades, while avoiding immune targeting of the glycosidase(s), which would otherwise eliminate its effectiveness over time.
  • the glycosidase composition and regimen is a cost effective treatment to reverse viral disease state or trajectory, and/or to transition to long term disease management.
  • the patient is a symptomatic AIDS patient
  • the glycosidase composition is provided with, or as an alternative to, ARV therapy, to reverse disease trajectory.
  • ARVs can be an effective antiviral treatment
  • ARV's have the adverse effect of suppressing the immune system, an effect harmful particularly for HIV or AIDS patients.
  • consistent immune modulation through the regimen described herein has the ability to ameliorate these side effects, while in the long term transitioning to the primary disease management.
  • the glycosidase composition after amelioration of the condition, allows transition to a cost effective regimen for long term disease management, which in some embodiments eliminates the need for chronic ARV therapy.
  • the regimen is generally effective for managing chronic viral infections such as HIV, HSV, EBV, HAV, HBV, HCV, HPV, adenovirus, and others.
  • the invention provides methods of treating and/or preventing an infectious disease.
  • the patient receives a regimen of the composition described herein, which provides and maintains effective immune stimulation over time, including as a lone or added protection during influenza season, or other ongoing infectious disease outbreak or epidemic.
  • the invention finds use with immunocompromised patients, including the elderly, children, the sick, hospitalized, and those with an immunodeficiency disorder (including genetic immunodeficiencies, drug-induced immunodeficiency, or due to infectious disease such as AIDS).
  • the composition and/or regimen acts as an adjuvant to enhance vaccine effectiveness, providing for more effective vaccination and/or longer duration of a vaccine's protective effect and in some embodiments, allows for vaccine dose sparing.
  • the invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising at least two of neuraminidase, galactosidase, N-acetylgalactosaminidase, fucosidase, glucosidase, N-acetylglucosaminidase, and mannosidase, and a pharmaceutically-acceptable excipient.
  • the composition may comprise neuraminidase and ⁇ -galactosidase.
  • the glycosidases may be present at, collectively, between about 10 ⁇ 3 mg to 10 ⁇ 8 mg.
  • the composition may be formulated for a variety of administration routes, including sublingual delivery.
  • the invention provides a convenient glycosidase dose applicator.
  • the applicator delivers a sufficient number of glycosidase doses for a non-acute regimen, such as for example, at least one month of doses for reversing viral disease trajectory or managing an infectious disease, or for preventing or mitigating infectious disease, or enhancing vaccine effectiveness.
  • the applicator maintains stability of the composition over the course of the regimen, protecting the composition from exposure to possible environmental contamination.
  • the glycosidase composition is stable over the length of time needed to administer the doses in accordance with a regimen described herein.
  • FIG. 1 shows the effect of neuraminidase and complement on the infectivity of Vero cells by HSV-1.
  • the indicated units of complement were incubated with neuraminidase and HSV-1. These mixtures were then added to Vero cell and incubated for 5 days. After cell fixation and staining, plaques were counted, with each plaque corresponding to one initial infectious viral particle. Infectivity is expressed as a percent of the virus control (virus incubated with cells and without complement or neuraminidase).
  • FIG. 2 shows the effect of neuraminidase and complement on the yield of infectious HSV-1 in Vero cells.
  • Vero cells were incubated with HSV-1. These infected cells were then incubated with mixtures containing the indicated units of complement and neuraminidase.
  • Controls consisted of infected cells without complement or neuraminidase (virus control). Following a 24-hour incubation, supernatants (containing released virus) from the cell cultures were tested for their ability to form plaques. Yield is expressed as a percent of the virus control.
  • FIG. 3 summarizes the effects of neuraminidase on the production of cytokines in vitro.
  • Cells appropriate for the production and measurement of the respective cytokines were incubated in the presence (experimental) or absence (control) of neuraminidase.
  • IL-2, IFN- ⁇ , and IFN- ⁇ total cellular RNA was extracted and hybridized with a cytokine-specific radioactive probe and counts per minute were determined.
  • For TNF- ⁇ optical densities were measured in a cell lytic assay. Values for each cytokine are expressed as the percent difference from the corresponding saline control. As indicated by the positive values, all four tested cytokines were stimulated in the presence of neuraminidase relative to controls.
  • the present invention provides compositions, methods and treatment regimens for non-acute immune enhancement, which finds application in the treatment of chronic infectious disease, including vaccine (e.g., adjuvant-free vaccine) enhancement.
  • vaccine e.g., adjuvant-free vaccine
  • the invention comprises administering an in vivo regimen of one or more glycosidase enzymes to a patient having an infectious disease.
  • the in vivo regimen stimulates immune signaling through removal of effective amounts of glycosides, notably sialic acids in some embodiments, from the surface of immune cells or other targets, and avoids excess removal of glycosides including sialic acid from normal cells.
  • the regimen allows persistent antigenic targeting of infected cells by the elicited immune cascade.
  • the regimen comprises enzymes active for removal of glycosides that are prevalent on virally infected cells, and which in various embodiments are terminal or penultimate glycosides.
  • the regimen comprises neuraminidase and a second glycosidase specific for the removal of a prevalent penultimate glycosyl residue, which can provide a synergistic treatment by avoiding, preventing, or slowing resialylation and/or re-capping of the glycosyl chains.
  • the administration regimen including as adjunct therapy and including embodiments that involve convenient patient dose monitoring, are as described in detail herein. According to this aspect, the effectiveness of the method does not critically rely on the identity of the infectious agent or the patient's unique biology, unlike many conventional therapies.
  • the invention in various embodiments provides for administering regimens of a glycosidase composition sufficient for immune stimulation, while also avoiding targeting of the glycosidase by the immune system, which might otherwise reduce or eliminate its effectiveness.
  • the invention further provides glycosidase dosing applicators for administering the regimens described herein.
  • the glycosidase composition comprises neuraminidase, which is an enzyme that hydrolyzes glycosidic linkages of terminal sialic acid residues on various glycoconjugates.
  • neuraminidase is an enzyme that hydrolyzes glycosidic linkages of terminal sialic acid residues on various glycoconjugates.
  • Neuraminidases are found in mammalian cells as well as various bacterial, fungal, and viral sources. By virtue of their terminal position on carbohydrate chains of cell membranes, sialic acids are key regulators of communication between cells and of immune recognition phenomena.
  • the neuraminidase is formulated, optionally with other glycosidases as described herein, and provided as a regimen of doses that allows it to function as a signaling cascade immunomodulator, coordinating the host's immune response to effectively combat infectious disease, including HIV and other viral and infectious agents, while not itself being (significantly) targeted by the immune system.
  • these properties of the composition are improved by proper formulation and/or delivery of the
  • the glycosidase therapy enhances immune function at least in part by complement activation.
  • HSV-1 Herpes simplex
  • in vitro studies show that incubating proper levels of neuraminidase and complement together significantly reduces both the infectivity of Vero cells by HSV-1 and the release of free virus from pre-infected cells relative to controls ( FIGS. 1 and 2 ).
  • the glycosidase therapy enhances immune function at least in part by cytokine stimulation, including, for example, Interleukin-2 (IL-2), interferon alpha (IFN- ⁇ ), interferon gamma (IFN- ⁇ ), tumor necrosis factor alpha (TNF- ⁇ ), Interleukin-4 (IL-4), and Interleukin-6).
  • cytokine stimulation including, for example, Interleukin-2 (IL-2), interferon alpha (IFN- ⁇ ), interferon gamma (IFN- ⁇ ), tumor necrosis factor alpha (TNF- ⁇ ), Interleukin-4 (IL-4), and Interleukin-6).
  • IL-2 Interleukin-2
  • IFN- ⁇ interferon alpha
  • IFN- ⁇ interferon gamma
  • TNF- ⁇ tumor necrosis factor alpha
  • IL-4 Interleukin-4
  • Interleukin-6 Interleukin-6
  • the glycosidase therapy enhances immune function at least in part by increasing efficiency of immune cell interactions.
  • the increased efficiency of these immune cell interactions in the presence of proper levels of neuraminidase, as well as other glycosidases, may be due to the removal of negatively charged sialic acid molecules, resulting in greater cell-to-cell contact.
  • the glycosidase composition and regimen enhances immune function by enhancing cell-mediated cytotoxicity and/or immune cell activation.
  • the glycosidase regimen functions at least in part by increasing exposure of infectious agent, such as virus (e.g., HIV, HSV, EBV, HCV, HPV, adenovirus) and viral infected cells to immune mechanisms,
  • infectious agent such as virus (e.g., HIV, HSV, EBV, HCV, HPV, adenovirus) and viral infected cells to immune mechanisms
  • virus e.g., HIV, HSV, EBV, HCV, HPV, adenovirus
  • removal of terminal sialic acid residues from gp120 of HIV or HIV-infected cells by neuraminidase may have other immune stimulating effects
  • Removing terminal sialic acid residues and/or other glycans from gp120/gp 160 of HIV either exposes hidden epitopes or alters the conformation of the glycoprotein such that the virus is more susceptible to attack by various components of the host's immune system.
  • the glycosidase formulation and regimen described herein tips the delicate
  • the glycosidase regimen which may comprise one or more of neuraminidase, ⁇ -galactosidase, ⁇ -mannidase, fucosidase (as well as other glycosidase enzymes, including those described herein) converts vitamin D binding protein (also known as group specific component, or Gc, to an effective macrophage activating factor in vivo, leading to activation of macrophages against the patient's infectious disease.
  • vitamin D binding protein also known as group specific component, or Gc
  • Gc group specific component
  • Vitamin D-binding protein also known as DBP
  • DBP Vitamin D-binding protein
  • DBP is an evolutionarily conserved glycoprotein, and is genetically polymorphic.
  • DBP has a relative molecular weight of about 52,000, and normally constitutes about 0.5% of the plasma protein.
  • the proper dose and regimen of glycosidase as described herein, can lead to effective, consistent, and chronic in vivo macrophage activation against the particular causative pathogen, including specific targeting of its antigenic state, which is crucial for pathogens that constantly change exposed epitopes.
  • in vivo administration of the glycosidase composition including convenient sublingual dosing, leads to effective macrophage activation against pathogens.
  • the invention provides a method for treating a patient having a chronic viral infection.
  • the method comprises administering a non-acute regimen of an immunotolerant and immune signaling glycosidase composition to the patient so as to treat, ameliorate, and/or manage said infection.
  • virus is one that integrates with the host genome (e.g., as provirus) or which can become latent, or otherwise escape immune surveillances as to be difficult or impossible to completely eliminate.
  • retroviruses e.g., HIV
  • herpes simplex viruses e.g., HSV-1 or HSV-2
  • hepatitis viruses e.g., HAV, HBV, HCV
  • adenovirus e.g., HIV-1 or HSV-2
  • the glycosidase composition and regimen described herein can take the place of antiviral chemotherapy or immunotherapy (e.g., cytokine or chemokine therapy, such as interferon, or monoclonal antibody treatment) for long term disease management, or can be combined with such therapies to improve outcome, that is, either combined simultaneously or in sequence.
  • antiviral chemotherapy or immunotherapy e.g., cytokine or chemokine therapy, such as interferon, or monoclonal antibody treatment
  • the patient is HIV positive, and in some embodiments is a symptomatic AIDS patient.
  • HIV infection is a worldwide problem, and various governments have made arrangements for their nationals to have access to antiretroviral drugs. In spite of these efforts, sometimes the drugs are not available because of poor communication and poor accessibility which results in missed doses by the patients. Also, quite often, patients are not able to tolerate these drugs.
  • the present invention provides alternatives to antiretroviral drugs in some embodiments, and in other embodiments, provides additional agents to reduce the need for long term use of antiretrovirals or other Chemotherapeutic or antiviral therapy.
  • the patient is not undergoing anti-retroviral therapy during the glycosidase treatment, for example, because the patient is unable to tolerate the anti-retroviral therapy, or such ARV therapy is not available to the patient.
  • the HIV may be any sub-type, such as HIV-1 or HIV-2.
  • the HIV is resistant to anti-retroviral chemotherapy, making the availability of an alternative therapy critical.
  • the glycosidase is administered or initiated after anti-retroviral therapy to manage chronic AIDS, thereby providing amore effective and cost-sensitive long-term disease management.
  • the glycosidase regimen may also be administered with anti-retroviral chemotherapy, for example to help reverse disease trajectory. Once disease trajectory is reversed, the glycosidase regimen may be optionally continued for at least one month, at least two months, at least four months, at least six months, or at least one year, or at least two years, or at least five years, or more, to provide a cost-effective management of the disease.
  • the frequency or daily dose may be adjusted for long term treatment as described herein.
  • the immune tolerated glycosidase signaling compensates for the loss of CD4 cells, while allowing the host time to recover and replenish its supply of these critical cells.
  • the glycosidase composition allows a subject on antiretroviral therapy to cease antiretroviral therapy for a period of time (e.g., about one to six months, or about one to four months, or about one to two months), thereby allowing the body to recuperate from the toxic effects of these drugs, while also providing cost advantages.
  • the regimen further allows the immune targeting of the virus and virus particles, as well as infected (e.g., lysogenic) cells preferentially over host cells.
  • this “cycle” of AVR and glycosidase treatment is repeated one or more times throughout therapy.
  • the administration of the glycosidase therapy with anti-retroviral therapy prevents some of the dampening of the immune system often exhibited by retroviral therapy.
  • Administration with other chemotherapeutic regimens, as described in this paragraph, for other viral infections can provide for the same or similar advantages.
  • Drugs designed to combat HIV have focused on the various stages of the viral life cycle. Some have been designed to block the virus' attachment to the CD4 T cell. Others have targeted the various enzymes involved in the assembly stages inside the cell; zidovudine (AZT), for example, is a well-known drug which functions to inhibit the activity of reverse transcriptase, an enzyme HIV uses to copy its RNA genome into DNA. Some of these synthetic drugs have slowed the progression to AIDS, but none have stopped it or eliminated the virus. A major problem has been the high mutation rate of HIV where drugs initially showing some promise become ineffective as the virus changes its form to one no longer recognized by the drug. The glycosidase regimen also targets HIV mutant derivatives allowing for enhancement of ARV as well as other possible chemotherapies, to reduce the propensity of mutant virus to circumvent the therapeutic.
  • anti-retroviral therapy refers to active agents (often cocktails of active agents) administered to HIV and AIDS patients. These include, for example, therapy comprising entry inhibitors (e.g., maraviroc and/or enfuvirtide), CCR5 receptor antagonists, reverse transcriptase inhibitors (e.g., zidovudine, didanosine, zalcitabine, stavudine, foscarnet, and/or lamivudine), protease inhibitors (e.g., ritonavir, darunavir, atazanavir, saquinavir), integrase inhibitors (e.g., raltegravir), maturation inhibitors alpha interferon, hevirimat, and/or becon).
  • entry inhibitors e.g., maraviroc and/or enfuvirtide
  • CCR5 receptor antagonists e.g., reverse transcriptase inhibitors (e.g., zidovudin
  • the retroviral therapy comprises a combination of two nucleoside-analogues and one non-nucleoside-analogue or protease inhibitor.
  • This three drug combination is commonly known as a “triple cocktail.”
  • Examples include COMBIVIR (zidovudine lamivudine), TRIZIVIR, (ahacavir+zidovudine+lamivudine), KALETRA (lopinavir+ritonavir), EPZICOM (abacavir+lamivudine), TRUVADA (tenofovir+emtricitabine), ATRIPLA (efavirenz+tenofovir+emtricitabine).
  • the anti-retroviral therapy comprises two nucleoside reverse transcriptase inhibitors and one non-nucleotide reverse transcriptase inhibitor.
  • the ARV therapy comprises Stavudine, Laminvudine, and/or Nevirapine.
  • the ARV therapy comprises Efavirenz.
  • Such therapies, if administered, are employed sparingly, or to reduce initial disease trajectory, before transitioning to long-term glycosidase treatment.
  • Such long term disease management with glycosidase therapy elicits an immune cascade against free virus, including mutant virus, and infected cells.
  • the regimen described herein in various embodiments is suitable for treating patients in various stages of AIDS.
  • the patient's CD4 count at the start of the regimen is less than about 500 cells per mm 3 .
  • the patient's CD4 count at the start of the regimen may be between about 200 and about 400.
  • the patient's CD4 count at the start of the regimen may be less than about 400, less than about 350, less than about 300, less than about 200, less than about 100, or less than about 50.
  • the CD4 count is less than 400
  • antiretroviral therapy is administered, either alone or with the glycosidase composition, and the glycosidase composition is used for long term care once the CD4 count is normalized (e.g., above about 400 or above about 500 or above about 800).
  • the patient may continue to have a below normal CD4 count, but the glycosidase composition helps the immune system function with this impairment.
  • the patient has a low CD4 count as described, and the glycosidase composition is administered without retroviral therapy, allowing the CD4 count to normalize in the long term.
  • the patient's vial load at the start of the regimen is above about 10,000 per ml.
  • the viral load at the start of the regimen may be at least about 25,000 per ml, at least about 40,000 per ml, at least about 50,000 per ml, at least about 75,000 per ml, at least about 100,000 per ml, at least about 500,000 per ml, at least about 1 million per ml, or at least about 5 million per ml.
  • the patient receives antiretroviral therapy to bring the viral load to less than about 50,000 per ml, or less than 10,000 per ml, or to undetectable, and then transitioning to glycosidase treatment as the long term care.
  • the combination of antiretroviral therapy and glycosidase allows for faster or more complete reduction in the viral load.
  • the benefit of glycosidase therapy is discernable at least in-part through an evaluation of the well being of the patient, including mood and mental state, appetite, energy level, secondary infections/complications, hair growth, and other symptoms of improving overall health.
  • Such benefits may or may not be discernable with conventional quantitation of AIDS treatment, such as CD4 level or viral titers.
  • the patient has a chronic viral infection selected from a herpes simplex virus infection (e.g., HSV-1 or HSV-2), Epstein-Barr Virus infection e.g., mononucleosis), cytomegalovirus infection, varicella zoster virus infection, hepatitis A, B, or C, adenovirus infection, or human papilloma virus infection.
  • a chronic viral infection selected from a herpes simplex virus infection (e.g., HSV-1 or HSV-2), Epstein-Barr Virus infection e.g., mononucleosis), cytomegalovirus infection, varicella zoster virus infection, hepatitis A, B, or C, adenovirus infection, or human papilloma virus infection.
  • the patient is diagnosed with chronic fatigue syndrome.
  • the glycosidase composition treats and/or prevents flare-ups of the disease and reduces viral load, and in some embodiments, takes the place of conventional antiviral strategies such as cytokine and/or chemokine therapies (e.g., interferon or interleukin) or small molecule antivirals (e.g., acyclovir, valaciclovir, famciclovir).
  • cytokine and/or chemokine therapies e.g., interferon or interleukin
  • small molecule antivirals e.g., acyclovir, valaciclovir, famciclovir
  • the glycosidase composition is administered for long term management, once the viral infection is under control by conventional therapy (e.g., interferon or small molecule virus inhibitor or monoclonal antibody therapeutic).
  • the regimen reduces or eliminates viral lesions such as cold sores, and/or prevents their reoccurrence.
  • the patient has an HSV or varicella zoster virus infection, and in some embodiments may have shingles.
  • the patient may not receive antiviral drugs such as acyclovir, valaciclovir and/or famciclovir, or in other embodiments, the glycosidase regimen is administered after the failure of conventional antivirals to ameliorate or eliminate the infection or symptoms thereof, or after conventional antivirals are rules by virtue of the patient's ability to tolerate these drugs.
  • the patient has hepatitis C infection, and receives interferon therapy.
  • the glycosidase regimen may be provided to replace ineffective INF therapy, for example, once the therapy loses effectiveness or is not tolerated by the patient.
  • the glycosidase regimen is provided alongside interferon therapy to boost its effectiveness.
  • the glycosidase regimen can facilitate the proper integration of interferon or antibody therapy for an effective immune response.
  • the invention provides methods of treating and/or preventing an infectious disease, other than a chronic viral infection described above.
  • the infectious disease is a persistent or recurrent bacterial infection, such as that associated with pneumonia, bronchitis, sinusitis, vaginitis, enteritis, colitis, sepsis, or urinary tract infection.
  • the regimen is further effective against persistent or recurrent ear, eye, nose and/or throat infection.
  • exemplary bacterial agents for which the invention may be effective include species of Mycobacterium (including tuberculosis), Pseudomonas (e.g., Psuedomonas aeruginosa , as may occur in association with cystic fibrosis), Haemophilus (e.g., Haemophilus influenzae ), Moraxella, Chlamydia, Neisseria, Streptococcus, Staphylococcus (including MRSA), Bordetella, Yersinia , and others.
  • the glycosidase regimen is administered after at least one round of antibiotic therapy has failed to ameliorate or eliminate the infection.
  • the glycosidase regimen is administered alongside antibiotic therapy, to enhance its effects, and reduce the potential for development of resistant bacteria.
  • antibiotics in these embodiments include an aminoglycoside, a carbapenum, a cephalosporin, a macrolide, a penicillin beta lactam), a quinolone (e.g., a fluoroquinolone such as ciprofioxacin), a sulfonamide, or a tetracycline, or combinations of the above.
  • the glycosidase regimen is effective against fungal or parasitic infections, which may be chronic, persistent, or recurring.
  • infections include Candidiasis (e.g., yeast vaginitis), malaria, trypanosomiasis, Aspergillus infection, toxoplasma, and Giardiasis.
  • the regimen may be administered after unsuccessful chemotherapy or antimicrobial treatment, or may be administered alongside the treatment to increase the rate of successful treatment, including elimination of the infectious agent or symptoms thereof in some embodiments.
  • the glycosidase regimen is administered for prevention of disease, especially where infectious disease is a particular risk, for example, during a Flu, SARS, or other outbreak.
  • the patient receives a non-acute regimen of the composition described herein, which provides effective glycosidase therapy for a period of time sufficient to span the period of outbreak.
  • the regimen can be administered to a patient at risk of contracting Flu, and the regimen provides for sustained immune stimulation throughout Flu season (e.g., at least two months, at least three months, at least four months, or at least six months).
  • This aspect of the invention is useful for other epidemics or outbreaks, including the protection of healthcare workers who are constantly exposed to highly contagious agents.
  • the invention finds use with immunocompromised patients, including the elderly, the young, the hospitalized, and patients with an immunodeficiency condition (e.g., resulting from AIDS, genetic disorder, or drug treatment), to boost immune function.
  • the glycosidase composition and/or regimen acts as a vaccine enhancer, providing for more effective vaccination, and/or longer duration of protective effect and in some embodiments, allows for vaccine dose sparing.
  • the vaccine is an adjuvant-free vaccine, with the glycosidase composition acting as the adjuvant.
  • the glycosidase composition and regimen may be initiated around the time of receiving a Flu or other vaccine (e.g., initiated within one week or three days or one day of receiving a vaccine), and the glycosidase regimen continued to lengthen the duration of the vaccine's protective effect and/or the level or duration of protective antibody titers.
  • the glycosidase regimen provides one or more glycosidase enzymes active for removal of one or more terminal and/or penultimate glycosyl groups on mammalian cells (e.g., immune cells and/or virally infected cells).
  • terminal and penultimate glycosyl groups include, for example, sialosyl, galactosyl, N-acetylgalactosamino, fucosyl, glucosyl, N-acetylglucosamino, and mannosyl residues.
  • the glycosidase regimen can include, in various embodiments, one or more of neuraminidase, galactosidase (e.g., ⁇ -Galactosidase), N-acetylgalactosaminidase, fucosidase, glucosidase, N-acetylgtucosaminidase, and mannosidase.
  • galactosidase e.g., ⁇ -Galactosidase
  • N-acetylgalactosaminidase e.g., fucosidase
  • fucosidase glucosidase
  • N-acetylgtucosaminidase e.g., glucosidase
  • mannosidase mannosidase
  • the glycosidase regimen comprises neuraminidase treatment.
  • the neuraminidase therapy may employ a neuraminidase or purified fraction haying neuraminidase (sialidase) activity, or an active portion or active derivative thereof.
  • the neuraminidase is microbial (e.g., bacterial, viral, parasitic, or fungal origin).
  • the neuraminidase is mammalian or plant.
  • the neuraminidase may be purified from food materials, including microbes that find use in foods, including baker's yeast and Lactococcus sp. and Lactobacillus sp.
  • the neuraminidase is bacterial.
  • the neuraminidase may be purified or isolated from its natural source, or may be recombinant or synthetic (e.g., chemically synthesized).
  • the neuraminidase is a ⁇ -Group B neuraminidase, also known as exo- ⁇ -sialidase, ⁇ -Group B, or acetyl Group B, which cleaves terminal sialic acid residues from carbohydrate moieties on the surfaces of host cells and virus.
  • the neuraminidase catalyzes the hydrolysis of ⁇ -2,3, ⁇ -2,6 and/or ⁇ -2,8 glycosidic linkages of terminal sialic acid residues in oligosaccharides, glycolipids and colominic acid.
  • the neuraminidase is an endo or exo sialidase, for example, catalyzing exo hydrolysis of ⁇ -(2 ⁇ 3), and/or ⁇ -(2 ⁇ 8) glycosidic linkages of terminal sialic acid residues, or catalyzing endo hydrolysis of (2 ⁇ 8)- ⁇ -sialosyl linkages in oligo- or poly(sialic) acid.
  • Exemplary neuraminidase agents include any of the well over 100 known neuraminidase enzymes, or active portion or derivative thereof,
  • the neuraminidase is an enzyme from one or more of Clostridium perfringes, Arthrobacter ureafaciens, Vibrio choterae, Salmonella typhimurium , or Streptococcus pneumoniae , or other whose activities are well characterized.
  • Such neuraminidase enzymes may be purified or isolated from its microbial source, or produced recombinantly or synthetically. See Cassidy J T, The Sialic Acids—VI. Purification and properties of sialidase from Clostridium perfringes, J. Biol.
  • the neuraminidase is at least 10% of the, protein component of the composition, at least 25% of the protein component of the composition, 50% of the protein component of the composition, or at least 75% of the total protein component, or at least 90% of the total protein component, or at least 95% of the total protein component, or at least 99% of the total protein component.
  • Exemplary amino acid sequences for neuraminidase proteins include those defined by GenBank accession numbers: EIA.17609.1, EIA17977.1, NP — 561469.1, CAA50436.1, ( Clostridium perfringes ), AAX22758.1, BAD66680.2 ( Arthrobacter ureafaciens ), AAW31751.2, AAA27546.1, AEA78761.1 ( Vibrio cholerae ), 2VWO_A ( Streptococcus Pneumoniae ), and AAL19864 ( Salmonella typhimurium ), which are each hereby incorporated by reference.
  • the neuraminidase may comprise an amino acid sequence having at least 70%, 80%, 90%, 95%, or more amino acid sequence identity to one or more of the amino acid sequences defined by EIA17609.1, EIA17977.1, NP — 561469.1, CAA50436.1, AAX22758.1, BAD66680.2, AAW31751.2, AAA27546.1, AEA78761.1, 2VWO_A, and AAL19864. Additional neuraminidase enzymes are described in U.S. Pat. No. 8,012,733, U.S. Pat. No. 6,916,916, U.S. Pat. No. 5,985,859, U.S. Pat. No. 5,830,748, and U.S. Pat. No. 4,071,408, which descriptions are hereby incorporated by reference in their entireties.
  • Suitable neuraminidases can be obtained from commercial sources.
  • Exemplary neuraminidase enzymes include Sigma Aldrich product numbers N2876, N3001, N5631, N2133 ( Clostridium perfringes ), N7885, N6514 ( Vibrio cholerae ), N3786, and N8271 ( Arthrobacter ureafaciens ).
  • neuraminidase enzymes may be guided by any of the known structures or studies, including those described by: Kim S et al., Features and applications of bacterial sialidases, Appl Microbial Biotechnol. 2011, 91(1):1-15; Crennell S J, et al., Crystal structure of a bacterial sialidase (from Salmonella typhimurium LT 2) shows the same fold as an influenza virus neuraminidase, Proc Natl Acad Sci USA 1993 90(21):9852-6; Chavas L M, Crystal structure of the human cytosolic sialidase Neu2: Evidence for the dynamic nature of substrate recognition, J Biol. Chem.
  • Neuraminidase activity can generally be described in terms of units (U), by determining the amount of sialic acid released from a suitable substrate, under defined conditions (e.g., pH 5.0 and 37° C.).
  • a suitable substrate e.g., NAN-lactose or bovine submaxillary mucin. See Warren L, J Biol. Chem. 234 1971 (1959).
  • the glycosidase regimen comprises galactosidase administration, which in some embodiments is ⁇ -galactosidase.
  • Galactosidase may be co-formulated with neuramindase or other enzyme in embodiments involving two or more glycosidasc enzymes.
  • Alpha- or Beta-galactosyl residues (including 1 ⁇ 3 linked and 1 ⁇ 4 linked) act as terminal glycosides on mammalian cells, including immune cells, and/or may be penultimate glycosyl residues, and may be linked to terminal sialic acids in some instances.
  • galactosidase e.g., ⁇ -galactosidase may be used independently according to the methods described herein, or may be used in conjunction with neuramindase or other glycosidase, which in some embodiments, prevents or slows resialylation or re-capping of glycosyl structures, thus rendering the regimen more effective, and supporting less frequent administrations and/or lower dosing.
  • Exemplary galactosidases are well known and commercially available.
  • ⁇ -Galactosidase may be obtained from Escherichia coil, Aspergillus oryzae, Kluyveromyces lactis , and Streptococcus pneumoniae , as well as other microbial (e.g., bacterial or fungal) and biological sources, including mammalian sources.
  • microbial e.g., bacterial or fungal
  • a suitable ⁇ -galactosidase may be obtained from Sigma-Aldrich catalogue number G5635.
  • the glycosidase regimen comprises N-acetylgalactosaminidase administration.
  • N-acetylgalactosaminidase may be co-formulated with neuramindase or other enzyme in embodiments involving two or more glycosidase enzymes.
  • N-acetylgalactosaminidase may be used independently according to the methods described herein, or may be used in conjunction with neuramindase or other glycosidase, which in some embodiments, prevents or slows resialylation or recapping of glycosyl chains.
  • N-acetylgalactosaminidases are well known and commercially available.
  • ⁇ -acetylgalactosaminidase may be obtained from Bacillus sp., as well as other microbial (e.g., bacterial or fungal) and biological sources, including mammalian sources.
  • microbial e.g., bacterial or fungal
  • a suitable ⁇ -N-acetylgalactosaminidase may be obtained from Sigma-Aldrich catalogue number A2464.
  • the glycosidase regimen comprises fucosidase administration, which is some embodiments is ⁇ -fucosidase.
  • Fucosidase may be co-formulated with neuramindase or other enzyme in embodiments involving two or more glycosidase enzymes (e.g., co-formulated with galactosidase or N-acetylgalactosaminidase).
  • Fucosyl residues (including ⁇ 1 ⁇ 2 linked, ⁇ 1 ⁇ 6 linked, and ⁇ 1 ⁇ 4 linked) act as terminal glycosides on mammalian cells, including immune cells, and/or may he penultimate glycosyl residues, and may be linked to terminal sialic acids in some instances.
  • fucosidase e.g., ⁇ -fucosidase
  • neuramindase may be used independently according to the methods described herein, or may be used in conjunction with neuramindase, which in some embodiments, prevents or slows resialylation re-capping of glycosyl chains, thus rendering the regimen more effective, and supporting less frequent administrations and/or lower dosing.
  • Exemplary fucosidase enzymes are well known and commercially available.
  • ⁇ -fucosidase may be obtained from Xanthomonas sp. (e.g., manihotis ), as well as other microbial (e.g., bacterial or fungal) and biological sources, including mammalian sources.
  • microbial e.g., bacterial or fungal
  • a suitable ⁇ -fucosidase may be obtained from Sigma-Aldrich catalogue numbers F3023 and F1924.
  • the glycosidase regimen comprises glucosidase administration, which in some embodiments is ⁇ -glucosidase.
  • Glucosidase may be co-formulated with neuramindase or other enzyme in embodiments involving two or more glycosidase enzymes (e.g., co-formulated with neuraminidase, mannosidase, or N-acetylglucosaminidase).
  • Glycosyl residues (including ⁇ 1 ⁇ 2 linked, ⁇ 1 ⁇ 3 linked, and ⁇ 1 ⁇ 4 linked) act as terminal glycosides on mammalian cells, including immune cells, and/or in some instances may be internal glycosyl residues.
  • glucosidase e.g., ⁇ -glucosidase
  • neuramindase mannosidase
  • mannosidase or other glycosidase
  • prevents or slows resialylation re-capping of glycosyl chains thus rendering the regimen more effective, and supporting less frequent administrations and/or lower dosing.
  • glucosidase enzymes are well known and commercially available.
  • ⁇ -glucosidase may be obtained from Sacchromyces cerevisiae, Aspergillas niger , or Bacillus stearothermophilus , as well as other microbial (e.g., bacterial or fungal) and biological sources (including food sources such as rice), and including mammalian sources,
  • microbial e.g., bacterial or fungal
  • biological sources including food sources such as rice
  • mammalian sources e.g., a suitable ⁇ -glucosidase may be obtained from Sigma-Aldrich catalogue numbers G5003, G0660, 70797, 49291, G9259, and G3651.
  • the glycosidase regimen comprises N-acetylglucosaminidase administration, which is some embodiments is ⁇ -N-acetylglucosaminidase.
  • N-acetylglucosaminidase may be co-formulated with neuramindase or other enzyme in embodiments involving two or more glycosidase enzymes (e.g., co-formulated with neuraminidase, mannosidase, and/or glucosidase).
  • N-acetylglucosamine residues act as terminal glycosides on mammalian cells, including immune cells, and/or in some instances may be penultimate or internal glycosyl residues.
  • Hakomori Aberant Glycosylation in Cancer Cell Membranes as Focused on Glycolipids: Overview and Perspectives, Cancer Research 45, 2405-2414 (1985); Dwek and Brooks, Harnesing Changes in Cellular Glycosylation in New Cancer Treatment Strategies, Current Cancer Drug Targets 4:425-442 (2004).
  • N-acetylglucosaminidase e.g., ⁇ -N-acetylglueosaminidase
  • neuramindase mannosidase
  • glucosidase which in some embodiments, prevents or slows resialylation, re-capping of glycosyl chains, thus rendering the regimen more effective, and supporting less frequent administrations and/or lower dosing.
  • N-acetylglucosaminidase enzymes are well known and commercially available.
  • ⁇ -N-acetylglucosaminidase may be obtained from Streptococcus pneumoniae and Canavalia ensiformis , as well as other microbial (e.g., bacterial or fungal) and biological sources (including food sources), and including mammalian sources.
  • microbial e.g., bacterial or fungal
  • biological sources including food sources
  • mammalian sources e.g., bacterial or fungal
  • a suitable ⁇ -N-acetylglucosaminidase may be obtained from Sigma-Aldrich catalogue numbers A2264 and A6803.
  • the glycosidase regimen comprises mannosidase administration, which in some embodiments is ⁇ -mannosidase.
  • Mannosidase may be co-formulated with neuramindase or other enzyme in embodiments involving two or more glycosidase enzymes (e.g., co-formulated with neuraminidase, glucosidase, and/or N-acetylglucosaminidase).
  • Mannosyl residues (including ⁇ 1 ⁇ 2 linked, ⁇ 1 ⁇ 3 linked, ⁇ 1 ⁇ 6 linked, ⁇ 1 ⁇ 4 linked, and others) act as terminal glycosides on mammalian cells, including immune cells, and/or in some instances may be penultimate or internal glycosyl residues.
  • mannosidase e.g., ⁇ -mannosidase
  • ⁇ -mannosidase may be used independently according to the methods described herein, or may be used in conjunction with neuramindase, glucosidase, and/or N-acetylglucosaminidase, which in some embodiments, prevents or slows resialylation re-capping of glycosyl chains, thus rendering the regimen more effective, and supporting less frequent administrations and/or lower dosing.
  • mannosidase enzymes are well known and commercially available.
  • mannosidase may be obtained from Canavalia enformis ( ⁇ ) or Helix pomatia ( ⁇ ), as well as other microbial (e.g., bacterial or fungal) and biological sources (including food sources), and including mammalian sources.
  • a suitable mannosidase may be obtained from Sigma-Aldrich catalogue numbers M7257 or M9400.
  • the glycosidase enzymes arc administered at a dose and frequency so as to exhibit a reduction in symptoms or pathology, without impacting normal cellular functions.
  • the dose and/or frequency of administration in some embodiments is a dose and/or frequency that does not cause prolonged joint discomfort or malaise (e.g., a general feeling of discomfort). Where joint or general discomfort is experienced by the patient, the patient may adjust the dose or frequency of administration until the discomfort subsides or normalizes.
  • the patient may skip one, two, or three days of dosing, and/or subtract one or two daily doses from the regimen, and/or increase the timing between doses, until the discomfort subsides or normalizes.
  • the patient finds the highest dose and/or frequency of administration that induces no prolonged joint discomfort, or minimal discomfort.
  • the amount of dose is controllable, for example using a metered dose applicator, the dose may be reduced but the schedule maintained.
  • each patient can tailor the dose as needed given the state of the patient's unique biology, disease or immune system condition, by finding the highest dose/frequency that does not induce prolonged joint discomfort or malaise.
  • the glycosidase formulation may be administered at less than approximately 10 ⁇ 2 or less than about 10 ⁇ 3 mg per dosage unit to a human or animal.
  • the glycosidase(s) are administered at between approximately 10 ⁇ 3 mg to 10 ⁇ 8 mg.
  • the dose of glycosidase is between approximately 10 ⁇ 3 mg and 10 ⁇ 7 mg, 10 ⁇ 3 mg and 10 ⁇ 6 mg, 10 ⁇ 3 mg and 10 ⁇ 5 mg, or is approximately 10 ⁇ 4 mg.
  • the total daily dose does not exceed about 10 ⁇ 3 mg per subject, or in some embodiments, does not exceed from about 5 ⁇ 10 ⁇ 3 to 10 4 mg.
  • patients exhibiting immune suppression may require higher doses within the range of 10 ⁇ 1 or 10 ⁇ 3 mg.
  • glycosidase(s) While certain glycosidases, including neuraminidases, can have a tendency to form homodimers (e.g., trimers, tetramers), in various embodiments the glycosidase(s) are formulated (e.g., diluted) to be present as a monomer and/or dimer, with substantially no higher aggregates as determinable by size exclusion chromatography (SEC).
  • SEC size exclusion chromatography
  • the glycosidase(s) may be formulated as an aqueous formulation, including for sublingual, nasal, or buccal delivery.
  • the aqueous formulation comprises saline.
  • the formulation has the ionic strength of from about 0.5 to about 2% saline, such as the ionic strength of about 0.9% saline.
  • the glycosidase(s) are formulated in normal saline (e.g., about 0.9% saline). Other conventional carriers for sublingual, nasal, or buccal delivery may also be employed.
  • the glycosidase(s) may be further formulated with a preservative, which may be an aromatic or phenolic preservative.
  • the preservative in some embodiments is phenol.
  • neuraminidase optionally with other glycosidases, is formulated in 0.05 to 0.5% phenol, or comparable amounts of similar acting preservative, for example.
  • the activity of the neuraminidase and potentially other glycosidases is increased by the presence of phenol, such as at least 0.2%, 0.3%, or 0.4% phenol.
  • the neuraminidase (e.g., Sigma Aldrich catalogue numbers N2876, N3001, N5631, N2133, N7885, N6514, N3786, N8271) is incubated in a solution containing from about 0.2% to about 1% phenol (e.g., from 0.2 to 0.6% phenol, or about 0.4% phenol), and then diluted to or brought to the final formulation, which may contain from 0.05% to about 0.2% phenol.
  • phenol e.g., from 0.2 to 0.6% phenol, or about 0.4% phenol
  • activation of the neuraminidase allows the active agent to be administered in lower doses to avoid immune targeting, while maintaining the proper level of activity.
  • neuraminidase and optionally with other glycosidase enzyme(s) can be mixed with 0.9% saline, and filter sterilized, and allowed to stand at room temperature for from 10 minutes to five hours (e.g., about 30 minutes to about three hours). After the incubation at room temperature, phenol saline is added to give a final phenol concentration of about 0.1% in 0.9% saline solution. The solution is stored at 4° C.
  • neuraminidase and optionally other glycosidase enzyme(s) is mixed with about 0.4% phenol saline.
  • This solution is filter sterilized, and allowed to stand at room temperature for from 5 minutes to about 5 hours (e.g., about 30 minutes, about one hour, or about three hours). After the incubation at room temperature, the final concentration is brought to about 0.1% phenol, 0.9% saline. The solution is stored at 4° C.
  • the glycosidase formulation may be administered by a variety of routes, including sublingual, nasal, port, subdermal, gavage, intraocular, intravenous, intramuscular, subcutaneous, transdermal, and buccal.
  • the glycosidases are administered sublingually.
  • the neuraminidase is held under the tongue for from about one to about five minutes, and preferably for about 3, about 4, or about 5 minutes. The patient should refrain from speaking during this time. The patient should not eat or drink within 15 minutes of administration.
  • regimens of glycosidase enzyme(s) are administered on average from 2 to 6 times per day for at least two weeks or at least one month, especially for the immune compromised or advanced cases.
  • the daily administrations should be substantially evenly spaced, but in various embodiments are spaced by about 15 minutes to 5 hours.
  • doses may be spaced by about 15 minutes, about 30 minutes, about 1 hour, about 2 hours, or about 3 hours.
  • the glycosidase formulation may be administered on average from 2 to 8 times per day for at least about two months, at least about four months, or at least about five months, or at least about six months.
  • the glycosidase formulation is administered about 2, about 3, or about 4 times per day over at least one month, two months, three months, four months, five months, or six months.
  • the glycosidase formulation is administered at a dose and frequency so as to be effective in reducing the infectious disease pathology or stimulating the immune system, without exhibiting substantial prolonged joint discomfort or malaise.
  • the patient adjusts the dose and/or frequency (e.g., skips one, two or three days of neuraminidase dosing, or reduces the daily dose by one or two administrations), until the discomfort subsides or normalizes.
  • the administration regimen is suspended during times of joint discomfot in some embodiments.
  • the first day of treatment may begin with about eight doses, the first three to five taken in the first one or two hours, with the remainder approximately evenly spaced throughout the day.
  • the patient may then be treated with about four doses per day, with periodic monitoring of the malignancy. Even where the malignancy is undetectable, the patient may remain on a regimen of 2 to 7 doses per day, as adjusted from time to time based on the appearance of joint discomfort or malaise.
  • the glycosidase regimen is an alternative to these conventional therapies.
  • the patient is subsequently treated chronically with about one dose per day, for at least about six months, or at least about one year, or at least about two years, or at least about five years, or more, or is selected or prescribed for such chronic treatment.
  • This subsequent chronic treatment in some embodiments is with the absence of chemotherapeutic or other therapy to reduce the likelihood of recurrence or disease progression.
  • Chronic glycosidase treatment for example, to prevent disease recurrence or relapse, may be administered 1 or 2 times per day.
  • the patient is instructed to monitor joint stiffness or malaise.
  • Such conditions suggest that glycosidase treatment should be adjusted.
  • the adjustment may include skipping one or two days or up to one week of dosing, or alternatively lowering the dose by one or two administrations per day, until the symptoms clear.
  • Other molecular assays could be used to the same effect, although joint stiffness or discomfort provides an ease of patient compliance.
  • the glycosidase dose can he easily adjusted per patient, and thus maintained chronically for optimal care.
  • a pharmaceutical composition comprising a delivery vehicle for administering a single glycosidase dose upon demand, and where the vehicle contains a full glycosidase regimen of at least 50 doses, or at least 100 doses, at least 150 doses, or at least 200 doses.
  • Each dose of glycosidase administered is an amount of up to about 10 ⁇ 2 mg glycosidase and pharmaceutically inert ingredients as already described.
  • the pharmaceutical composition may comprise at least two of neuraminidase, galactosidase, N-acetylgalactosaminidase, fucosidase, glucosidase, N-acetylgtucosaminidase, and mannosidase, and a pharmaceutically-acceptable excipient.
  • the composition may comprise neuraminidase and ⁇ -galactosidase.
  • the glycosidases may be present at, collectively, between about 10 ⁇ 3 mg to 10 ⁇ 8 mg, or according to the doses disclosed above.
  • the composition may be formulated for a variety of administration routes as disclosed herein, including sublingual delivery.
  • the treatment regimen involves the partitional administration of an amount not to exceed approximately 10 ⁇ 3 mg of glycosidase, although, in certain cases, the total amount of glycosidase administered in any one day may exceed this limit.
  • the glycosidase formulation can be administered in a variety of routes and forms.
  • the glycosidase can be administered as a solid where the enzymes are embedded or admixed in a biodegradable or bioerodable matrix.
  • the matrix can be a time release matrix, These matrices are well known to those of ordinary skill in the art.
  • the glycosidase can be administered by injection or by sublingual route.
  • the vehicle is an aqueous solution that is contained within an inert container.
  • the composition is in the form of a suppository.
  • the liquid form of the composition can be injected subcutaneously, intramuscularly or intravenously.
  • composition can be administered through the mucosal membranes such as nasal membranes.
  • the glycosidase composition is administered via a drug applicator, the applicator comprising at least 100 doses of the composition, or at least 150 doses, or at least 200 doses.
  • the applicator is for sublingual, nasal, transdermal, time release sub-dermal, intraocular, gavage, port, subcutaneous, oral, or buccal. delivery.
  • the applicator is for sublingual delivery.
  • the applicator delivers a metered dose, that can be adjusted by the patient as needed.
  • the applicator preferably dispenses doses in a manner that maintains aseptic conditions of the remaining doses.
  • the applicator can be any of those that are described in U.S. Pat. Nos. 4,830,284; 4,565,302; 5,011,046; 5,147,087; 5,893,484; 6,877,672; 6,886,556, and 7,201,296, which are each hereby incorporated by reference in their entireties.
  • the applicator can be an atomizing or dosing pump, which can ensure that the medium present in the area between the pump cylinder and the discharge opening does not dry or is not otherwise altered by ambient influences. See U.S. Pat. No.
  • the applicator employs a 0.2 ⁇ m filter to maintain aseptic contents. Additionally, the applicator can dispense doses in a single-stroke discharge. Such applicators are described in U.S. Pat. No. 5,893,484, which is hereby incorporated by reference.
  • the applicator may be configured for nasal delivery, dermal delivery, throat delivery, or sublingual delivery.
  • the applicator allows for an actuatable dosing mechanism, which permits monitoring of precise doses and therefore largely eliminates incorrect dosing with respect to the number of doses and/or the duration dosing. See U.S. Pat. No. 4,565,302, which is hereby incorporated by reference.
  • the applicator delivers a dose in from 50 to 100 ⁇ l, such as the applicators described in, for example, U.S. Pat. No. 6,886,556, which is hereby incorporated by reference.
  • HSV-1 as model virus, in vitro studies were initiated to determine the effect of neuraminidase and complement on viral infectivity and release from Vero cells.
  • combinations of complement, virus and neuraminidase were incubated together, then added to Vero cells and incubated further. Following cell fixation and staining, the virus-forming plaques in the cells were counted. Results showed that incubating the virus with neuraminidase and complement together significantly (70-80%) reduced the virus' infectivity of Vero cells relative to controls ( FIG. 1 ).
  • Vero cells were first infected with HSV-1. These cells were then incubated with various concentrations of complement and neuraminidase. Supernatants from the cell cultures were tested for plaque-forming ability, indicative of virus released from the cells. Results showed that, in the presence of both neuraminidase and complement, the release of free virus was greatly decreased (70-80%) relative to controls ( FIG. 2 ). These results indicate that a. combination of neuraminidase and complement effectively reduced both the infectivity of Vero cells by HSV-1 and the release of free virus from pre-infected cells.
  • Cytokines are chemical messengers secreted by activated lymphocytes in response to infection. Any given cytokine, either alone or in combination with other cytokines, can have multiple effects on immune function.
  • Interleukin-2 (IL-2), interferon alpha (IFN- ⁇ ), interferon gamma (IFN- ⁇ and tumor necrosis factor alpha (TNF- ⁇ ) are important cytokines involved in host defense against viruses, IL-2, TNF- ⁇ and IFN- ⁇ are produced by the T-helper (TH1) subset of T cells and are therefore associated with the inflammatory process.
  • IL-2 and IFN- ⁇ together activate macrophages, which are important immune cells that engulf and digest pathogens (phagocytosis), and also serve as antigen-presenting cells to T lymphocytes.
  • IL-2 and IFN- ⁇ also enhance the cytotoxicity of natural killer (NK) cells in clearing virally-infected cells.
  • IFN- ⁇ also enhances the expression of major histocompatibility complex (MHC) class I and II molecules on antigen-presenting cells, thereby inducing CD4+ and CD8+ cytolytic cells involved in viral clearance.
  • MHC major histocompatibility complex
  • IFN- ⁇ also combines with TNF- ⁇ to stimulate NK cells. IFN- ⁇ inhibits viral replication by blocking the transcription of early viral proteins.
  • FIG. 3 summarizes the results obtained when cells treated with neuraminidase were tested for the production of IL-2, IFN- ⁇ , IFN- ⁇ , and TNF- ⁇ .
  • the production of all four cytokines was stimulated using the therapeutic dose of neuraminidase relative to the respective controls. It is reasonable to expect that the functions of these cytokines discussed above could be enhanced by neuraminidase in an in vivo system, and would have anti-viral effects in patients.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Organic Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Virology (AREA)
  • Epidemiology (AREA)
  • Communicable Diseases (AREA)
  • Immunology (AREA)
  • Oncology (AREA)
  • Pulmonology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Genetics & Genomics (AREA)
  • General Engineering & Computer Science (AREA)
  • Nutrition Science (AREA)
  • Physiology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biotechnology (AREA)
  • Otolaryngology (AREA)
  • Hematology (AREA)
  • AIDS & HIV (AREA)
  • Urology & Nephrology (AREA)
  • Ophthalmology & Optometry (AREA)
US14/431,249 2012-09-28 2013-09-26 Glycosidase regimen for treatment of infectious disease Abandoned US20150238580A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/431,249 US20150238580A1 (en) 2012-09-28 2013-09-26 Glycosidase regimen for treatment of infectious disease

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201261707252P 2012-09-28 2012-09-28
PCT/US2013/061966 WO2014052621A1 (en) 2012-09-28 2013-09-26 Glycosidase regimen for treatment of infectious disease
US14/431,249 US20150238580A1 (en) 2012-09-28 2013-09-26 Glycosidase regimen for treatment of infectious disease

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2013/061966 A-371-Of-International WO2014052621A1 (en) 2012-09-28 2013-09-26 Glycosidase regimen for treatment of infectious disease

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/204,398 Continuation US10675336B2 (en) 2012-09-28 2018-11-29 Glycosidase regimen for the treatment of chronic viral infection

Publications (1)

Publication Number Publication Date
US20150238580A1 true US20150238580A1 (en) 2015-08-27

Family

ID=50388969

Family Applications (3)

Application Number Title Priority Date Filing Date
US14/431,249 Abandoned US20150238580A1 (en) 2012-09-28 2013-09-26 Glycosidase regimen for treatment of infectious disease
US16/204,398 Expired - Fee Related US10675336B2 (en) 2012-09-28 2018-11-29 Glycosidase regimen for the treatment of chronic viral infection
US16/865,652 Abandoned US20200261557A1 (en) 2012-09-28 2020-05-04 Glycosidase regimen for treatment of infectious disease

Family Applications After (2)

Application Number Title Priority Date Filing Date
US16/204,398 Expired - Fee Related US10675336B2 (en) 2012-09-28 2018-11-29 Glycosidase regimen for the treatment of chronic viral infection
US16/865,652 Abandoned US20200261557A1 (en) 2012-09-28 2020-05-04 Glycosidase regimen for treatment of infectious disease

Country Status (11)

Country Link
US (3) US20150238580A1 (ja)
EP (2) EP2900259B1 (ja)
JP (1) JP6673696B2 (ja)
KR (2) KR20210019605A (ja)
CN (2) CN104902926A (ja)
AU (2) AU2013323472B2 (ja)
CA (1) CA2886474A1 (ja)
DK (1) DK2900259T3 (ja)
ES (1) ES2805795T3 (ja)
MX (2) MX2015004009A (ja)
WO (1) WO2014052621A1 (ja)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20190048558A1 (en) * 2017-08-08 2019-02-14 Entro Industries, Inc. Automatic walking for a load transporting apparatus
WO2022040090A1 (en) * 2020-08-17 2022-02-24 The Regents Of The University Of California Application of microbial glycosidase as an anti-viral therapeutic, prognostic, and diagnostic

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN106102776B (zh) 2014-01-24 2020-02-14 圣安德鲁斯大学董事会 免疫调节化合物
GB201616007D0 (en) 2016-09-20 2016-11-02 Univ Court Of The Univ Of St Andrews The Novel Adjuvants
GB201616009D0 (en) 2016-09-20 2016-11-02 Univ Court Of The Univ Of St Andrews The Treatment and/or prevention of sepsis
GB201616006D0 (en) 2016-09-20 2016-11-02 Univ Court Of The Univ Of St Andrews The Cell modulation
CN113663053A (zh) * 2021-08-20 2021-11-19 广西壮族自治区兽医研究所 Ifi6蛋白或调控ifi6蛋白基因表达的物质在制备禽呼肠孤病毒抑制剂中的应用

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5558863A (en) * 1991-04-09 1996-09-24 Molecular Rx, Inc. Method for treatment of herpes virus infections
WO2013038997A1 (ja) * 2011-09-14 2013-03-21 医療法人再生未来 医薬組成物およびその製造方法

Family Cites Families (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE2620649C3 (de) * 1976-05-11 1980-11-06 Behringwerke Ag, 3550 Marburg Immunologisches Adjuvans
US4071408A (en) 1976-11-01 1978-01-31 Research Corporation Neuraminidase
DE3302160A1 (de) 1983-01-22 1984-07-26 Ing. Erich Pfeiffer GmbH & Co KG, 7760 Radolfzell Betaetigbare dosiereinrichtung
DE3315334A1 (de) 1983-04-28 1984-10-31 Pfeiffer Erich Gmbh & Co Kg Zerstaeuber- oder dosierpumpe
DE3715301A1 (de) 1987-05-08 1988-11-24 Pfeiffer Erich Gmbh & Co Kg Austragvorrichtung fuer medien
DE3722469A1 (de) 1987-07-08 1989-01-19 Pfeiffer Erich Gmbh & Co Kg Handbetaetigbare austragvorrichtung fuer medien
US5326749A (en) 1989-11-20 1994-07-05 Nobuto Yamamoto Macrophage activating factor from vitamin D binding protein
US5177002A (en) * 1989-11-20 1993-01-05 Nobuto Yamamoto In vitro enzymatic conversion of glycosylated human vitamin D binding protein to a potent macrophage activating factor
GB9018061D0 (en) * 1990-08-17 1990-10-03 Wellcome Found Compositions for vaccines
IT1252228B (it) 1991-12-17 1995-06-05 T Associated Bio Technologies Procedimento per la produzione di neuraminidasi
US6562588B2 (en) 1993-05-17 2003-05-13 Genentech, Inc. Sialidase and recombinant cell lines
US5985859A (en) 1994-04-14 1999-11-16 The University Of Alabama Methods of inhibiting bacterial sialidase
DE19525734A1 (de) 1995-07-14 1997-01-16 Pfeiffer Erich Gmbh & Co Kg Austragvorrichtung für fließfähige Medien, insbesondere für den Austrag in nur einem Hub
AU7126998A (en) * 1997-04-16 1998-11-11 Connaught Laboratories Inc. Anti-influenza compositions supplemented with neuraminidase
US7157083B2 (en) * 1998-04-17 2007-01-02 Surrogate Pharmaceutical Pathways, Llc Compositions and methods for treating retroviral infections
DE60128793T2 (de) 2000-04-07 2008-02-07 Signal Coordinating Therapy Inc. Verfahren und Zusammensetzung zur Behandlung von Neoplasmen
AU2001288216A1 (en) * 2000-07-03 2002-01-14 Fisher Scientific Company L.L.C. System and method for web-based electronic buying system
CU23235A1 (es) * 2001-02-28 2007-09-26 Ct Ingenieria Genetica Biotech POXVIRUS RECOMBINANTES PARA PROTEINAS QUIMéRICAS DEL VIRUS DE LA INMUNODEFICIENCIA HUMANA Y SU APLICACION EN LA TERAPéUTICA Y LA PREVENCION DEL SIDA
DE10146815B4 (de) 2001-09-18 2005-05-04 Ing. Erich Pfeiffer Gmbh Spender für Medien
WO2003026803A1 (de) 2001-09-21 2003-04-03 Ing. Erich Pfeiffer Gmbh Dosiervorrichtung mit einer pumpvorrichtung
DE10164452A1 (de) 2001-12-21 2003-07-03 Pfeiffer Erich Gmbh & Co Kg Spender für Medien
CN1473847A (zh) * 2002-08-06 2004-02-11 中国科学院福建物质结构研究所 八棱丝瓜蛋白1的糖苷酶活性及其应用
CN1473848A (zh) * 2002-08-06 2004-02-11 中国科学院福建物质结构研究所 八棱丝瓜蛋白2的抗肿瘤活性及其应用
WO2004062565A2 (en) * 2003-01-08 2004-07-29 E. Nechmad Ltd. Device and method for vaccination
CA2558829C (en) * 2004-04-02 2014-03-11 Zymogenetics, Inc. Methods for treating viral infection using il-28 and il-29 cysteine mutants
US7892752B2 (en) 2005-04-26 2011-02-22 Dwek Raymond A Glycosylation markers for cancer diagnosing and monitoring
EP2084527A4 (en) 2006-11-02 2011-07-27 Seattle Genetics Inc METHOD FOR TREATING NEOPLASIA, AUTOIMMUNE AND INFLAMMATORY DISEASES
JP2010516784A (ja) * 2007-01-29 2010-05-20 ユニバーシティ・オブ・オックスフォード 糖免疫原
CA2676686C (en) 2007-02-20 2015-12-15 Dsm Ip Assets B.V. Novel sialidase
EP2155628A2 (de) 2007-04-24 2010-02-24 CeramTec AG Bauteil mit einem metallisierten keramikkörper
CN101214252B (zh) * 2008-01-08 2011-03-16 山东大学 一种抗病毒抗菌药物组合物及其制备方法
KR20170124619A (ko) * 2009-03-27 2017-11-10 아카데미아 시니카 바이러스에 대한 면역을 위한 방법 및 조성물
WO2014113641A1 (en) 2013-01-18 2014-07-24 Kline Ellis Selective glycosidase regimen for immune programming and treatment of cancer

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5558863A (en) * 1991-04-09 1996-09-24 Molecular Rx, Inc. Method for treatment of herpes virus infections
WO2013038997A1 (ja) * 2011-09-14 2013-03-21 医療法人再生未来 医薬組成物およびその製造方法
US8747919B2 (en) * 2011-09-14 2014-06-10 Saisei Mirai Clinic Pharmaceutical composition and method of preparing same

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Homer et al. (J. Med. Microbiol., 1994, 41:184-190 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20190048558A1 (en) * 2017-08-08 2019-02-14 Entro Industries, Inc. Automatic walking for a load transporting apparatus
WO2022040090A1 (en) * 2020-08-17 2022-02-24 The Regents Of The University Of California Application of microbial glycosidase as an anti-viral therapeutic, prognostic, and diagnostic

Also Published As

Publication number Publication date
CN104902926A (zh) 2015-09-09
AU2018203958A1 (en) 2018-06-21
AU2013323472B2 (en) 2018-07-05
DK2900259T3 (da) 2020-07-13
CA2886474A1 (en) 2014-04-03
AU2013323472A1 (en) 2015-04-16
MX2015004009A (es) 2015-12-09
EP2900259A4 (en) 2016-07-13
ES2805795T3 (es) 2021-02-15
JP6673696B2 (ja) 2020-03-25
JP2015532286A (ja) 2015-11-09
EP2900259B1 (en) 2020-04-15
US20190091305A1 (en) 2019-03-28
KR20210019605A (ko) 2021-02-22
KR20150063079A (ko) 2015-06-08
EP2900259A1 (en) 2015-08-05
MX2020011107A (es) 2020-11-11
AU2018203958B2 (en) 2020-02-20
CN112755178A (zh) 2021-05-07
WO2014052621A1 (en) 2014-04-03
US10675336B2 (en) 2020-06-09
US20200261557A1 (en) 2020-08-20
EP3741387A1 (en) 2020-11-25

Similar Documents

Publication Publication Date Title
US20200261557A1 (en) Glycosidase regimen for treatment of infectious disease
JP3246918B2 (ja) 感染症の治療のためのIL―12およびIFNαの使用
ES2767304T3 (es) Utilización de compuestos de tiazolida para la prevención y el tratamiento de enfermedades víricas, cáncer y enfermedades causadas por infecciones intracelulares
WO2007143934A1 (fr) Composition pharmaceutique destinée à la prophylaxie et au traitement d'une infection par le vih et son utilisation
JP2023123440A (ja) 合成リジンアナログ及び模倣物の抗ウイルス用途のための方法及び組成物
Wang et al. Reactive oxygen species associated immunoregulation post influenza virus infection
EP4121092B1 (en) Hybrid interferons for treating viral infections
WO2021142320A1 (en) Methods of using andrographolide, oridonin and isoliquiritigenin and derivatives thereof
EP2259799B1 (en) Combination of immunomodulator and anti-pathogenic agent
US10765696B2 (en) Compositions of and methods of treatment with zwitterionic polysaccharide
Pluta et al. The use of interferon in medicine
US20130178466A1 (en) Treatment of Microbial Infections
EP0579765B1 (en) Method and composition for the treatment of herpes related disorders
US20060063713A1 (en) Methods for restoring immune balance for the treatment of T-cell mediated diseases
CN112972453B (zh) MDL-800在制备抑制SARS-CoV-2病毒易感性药物中的应用
AU2017221785B2 (en) Use of thiazolide compounds for the prevention and treatment of viral diseases, cancer and diseases caused by intracellular infections
WO2004002511A1 (ja) 抗hiv剤
IE83322B1 (en) Method and composition for the treatment of herpes related disorders
EP1446142A1 (en) Potentiating the therapeutic effects of interferons

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION